Next Article in Journal
A Hypovirulence-Associated Partitivirus and Re-Examination of Horizontal Gene Transfer Between Partitiviruses and Cellular Organisms
Previous Article in Journal
Challenges and Opportunities for Post-COVID Pulmonary Disease: A Focused Review of Immunomodulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ferroptosis in Cancer: Mechanism and Therapeutic Potential

1
Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India
2
Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(8), 3852; https://doi.org/10.3390/ijms26083852
Submission received: 11 February 2025 / Revised: 11 April 2025 / Accepted: 15 April 2025 / Published: 18 April 2025
(This article belongs to the Section Molecular Oncology)

Abstract

:
Cancer drug resistance occurs when cancer cells evade cell death following treatment with chemotherapy, radiation therapy, and targeted therapies. This resistance is often linked to the reprogramming of programmed cell death (PCD) pathways, allowing cancer cells to survive drug-induced stress. However, certain anticancer therapies, when combined with specific agents or inhibitors, can induce ferroptosis—a form of cell death driven by iron-dependent lipid peroxidation. Currently, extensive preclinical and clinical research is underway to investigate the molecular, cellular, and tissue-specific mechanisms underlying ferroptosis, with the goal of identifying strategies to overcome drug resistance in cancers unresponsive to conventional PCD pathways. By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes in patients with cancer. This short review aims to enhance the understanding of ferroptosis inducers in cancer therapy and stimulate further research into ferroptosis-based approaches for more effective clinical cancer treatment.

1. Introduction

Tumor heterogeneity (TMH) refers to the diversity of tumor cells within a tumor or among different tumors. Even within a particular tumor, the cells differ in morphology, proliferation, metastatic potential, gene expression patterns, and metabolic requirements. TMH exists at the molecular, cellular, and tissue levels and is responsible for survival, cell death, and resistance to cancer drugs [1,2]. Resistance to conventional anticancer drugs is attributed to the complex and evolving nature of tumors [3,4]. The emergence of drug resistance in cancer cells provides distinctive advantages, such as proliferation, survival, evasion of immune destruction, and plasticity [3,4].
Ferroptosis is a distinct form of regulated cell death that involves cellular lipid peroxidation and iron metabolism [3,4,5]. Lipid metabolic pathways that generate lipid peroxides, when combined with excess iron ions, trigger ferroptosis type of cell death. Lipid peroxides accumulate when their removal mechanisms, such as the glutathione-dependent antioxidant system (GPX4 enzyme), are impaired. This has potential therapeutic implications for cancer treatment [6,7]. Ferroptosis is a form of caspase-independent programmed cell death driven by the mitochondrial and membrane-associated accumulation of reactive oxygen species and lipid peroxides [8,9,10,11]. Understanding the molecular mechanisms of ferroptosis, including iron metabolism, lipid peroxidation, and antioxidant defenses, could help develop combination therapies with existing cancer treatments [12,13,14,15,16,17].
In this review, we discuss ferroptosis from the perspective of its use as a novel strategy for combinatorial anticancer drug development at both preclinical and clinical levels. We hope that this discussion will stimulate further research into ferroptosis-based approaches for more effective cancer treatment in clinical practice.

2. Types of Drug-Induced Cell Death

The acquisition of functional capacities by human cells during the transition from normal to neoplastic development stages is attributed to many critical characteristics known as tumor hallmarks [18,19]. Among the key hallmarks of tumor, the interdependence and crosstalk between cancer cell drug resistance and metabolic reprogramming have been extensively studied and determined to provide several advantages to cancer cells in a drug-induced environment [20,21,22,23]. Therefore, therapeutic management of deregulated cellular metabolism, such as lipid metabolism [24,25], oxidative stress [26], iron-induced ROS formation [27,28], and overcoming resistance, is being explored at preclinical and clinical levels [29,30].

2.1. Apoptosis

Cancer therapies target apoptosis, a programmed form of cell death [31]. However, mutations in apoptotic pathways promote treatment resistance [32,33], allowing cancer cells to survive and proliferate [34,35]. Alternative cell death mechanisms like necrosis [36], senescence [37], autophagy [38], and mitotic catastrophe [39] enable further evasion, presenting potential drug targets [40,41,42,43]. Mutations in caspases [44], p53 [45], and Bcl-2 proteins [46] weaken apoptotic control, driving unregulated growth. Cancer cells enhance their survival by upregulating anti-apoptotic proteins, mutating pro-apoptotic genes, and altering pathways like PI3K/Akt and NF-κB [47,48]. Apoptosis maintains tissue homeostasis by removing damaged cells [49]; however, frequent mutations in p53 [50,51] and overexpression of Bcl-2 proteins [52] inhibit this process. Caspases, which are essential for apoptosis, are often suppressed in cancer cells [53,54]. Traditional therapies induce apoptosis through intrinsic and extrinsic pathways [55], involving caspase activation [56] and cellular changes like chromatin condensation [57]. While apoptosis relies on a balance between pro- and anti-apoptotic factors [58], mutations in TP53 [59] often disrupt this process. Many chemotherapeutic and targeted drugs function by inducing apoptosis [60].

2.2. Autophagy

Autophagy is a process in which cells digest and recycle their constituents [61]. Autophagy generally promotes cell survival by supplying nutrients during stress and maintaining homeostasis [62]. However, autophagy can also cause cell death, especially in apoptosis-resistant cancer cells [63]. Certain cancer therapies modulate autophagy to facilitate death in cancer cells [64]. For instance, a few mTOR inhibitors have been shown to induce autophagy, resulting in tumor cell death, especially when used in combination with other modalities of treatment [65]. Autophagy can act as a survival mechanism and as a form of cell death [66]. Autophagy has been reported to be regulated by many signaling pathways, including PI3K/Akt/mTOR [67], and often results from responses to metabolic and therapeutic stressors [68]. Pharmacological agents are known to modulate autophagy and cause cell death in apoptosis-resistant cancer cells [69].

2.3. Necrosis

Necrosis is uncontrolled cell death or a form of accidental cell death caused by extreme stress or damage, where the cell membrane bursts, leading to inflammation [70]. Although necrosis is generally not targeted in cancer therapy, some treatments may cause unintended necrosis within tumors, resulting in self-destruction [71]. The process of necrosis normally occurs due to overwhelming cellular stress that negatively affects mitochondrial function, resulting in energy depletion and activation of proteases, leading to cellular membrane rupture [72]. Programmed necrosis may be induced via mediators involving RIP kinases [73], PARP [74], and TLR-4 signaling pathways [75], particularly those mediated in response to stimuli, including graphene-based materials [76]. The induction of both apoptosis and autophagy can prevent necrosis, demonstrating that cell death pathways are delicately balanced, where a major part does not prevent but induces counter-cell-death pathways [77].

2.4. Necroptosis

Necroptosis, or regulated necrosis, is a type of cell death characterized by the disintegration of the plasma membrane and oncosis (swelling of subcellular organelles) without displaying the unique features of apoptosis or autophagy [78]. It is induced by the kinase activity of receptor-interacting serine/threonine kinase protein (RIPK) 1, thereby forming complex IIB and causing cell necroptosis; RIPK 1 and RIPK 3 are responsible for the phosphorylation of mixed-lineage kinase-like (MLKL) [79,80]. The activation of cell surface death receptors (such as FasRs, TNFR1, IFN receptors, and TLRs) and RNA- and DNA-detecting molecules in cells initiates the necroptotic process [78,79,80]. Studies on drug-induced cell death have revealed the complex mechanisms through which apoptosis, autophagy, and necrosis are regulated and connected [81]. In addition to these known cell death pathways, ferroptosis offers a way to bypass these traditional forms of cell death, utilizing iron-catalyzed lipid peroxidation as its killing mechanism. These are vital for understanding cancer therapy and the development of new therapeutic approaches.

2.5. Ferroptosis

Ferroptosis is an iron-dependent, non-apoptotic form of regulated cell death driven by lipid peroxidation [82]. First identified by Dixon et al. [6], it differs from apoptosis, necrosis, and autophagy in terms of structure and function and is gaining therapeutic interest [80,81,82,83,84,85]. Cell fate is influenced by oxidative stress, which is a key factor in metabolism and survival [86,87]. Various environmental and genetic factors, including heat [88], radiation [89], metabolism [90], redox homeostasis [91], immune surveillance [92], and oncogenic signaling [93] contribute to oxidative stress, triggering ferroptosis through unchecked lipid peroxidation [94]. Ferroptosis is characterized by mitochondrial shrinkage, outer membrane damage, increased membrane density, and reduced NADH levels, without chromatin condensation [95,96,97,98,99,100]. Unlike apoptosis, it does not require caspase activation [101] and is unaffected by necrosis regulators RIP1/RIP3 and Cyclophilin D [102] or autophagy inhibition by 3-MA [103], confirming its distinct nature [104]. Identified as a potential cancer therapy, ferroptosis can target resistant cells through iron-catalyzed lipid peroxidation [104,105,106]. It results from iron overload [107] and metabolic imbalance [108] and is regulated by genes such as GPX4 [109] and ACSL4 [110], which control lipid metabolism and antioxidant defense [111].

2.6. Ferroptosis: Molecular Mechanism

Ferroptosis is an iron-dependent, intrinsically regulated cell death that is distinct from apoptosis and necrosis based on iron dependence and the accumulation of lipid peroxides [112]. These processes are complex and interwoven with iron metabolism, lipid peroxidation, and antioxidant defense mechanisms [113]. Ferroptosis requires labile iron, which catalyzes the formation of lipid hydroperoxides, mainly through the action of lipoxygenases [114]. The antioxidant enzyme GPX4 utilizes GSH to regulate ferroptosis by reducing lipid hydroperoxides [115]. When homeostasis is disturbed or antioxidant defenses are compromised, lipid hydroperoxides accumulate, causing cell death [116]. Therefore, it is important to understand the pathways involved in ferroptosis, as therapeutics may be applied to diseases associated with ferroptosis [117].
Ferroptosis, a form of regulated cell death, is characterized by the formation of ●OH generated from the reaction of H2O2 with Fe2+ (the Fenton reaction) [118]. Ferroptosis is triggered by lipid peroxidation, which is driven by hydroxyl radicals (●OH) reacting with polyunsaturated fatty acids (PUFAs) in the cell membrane. This process generates lipid hydroperoxides, which accumulate if not neutralized by antioxidant systems, such as glutathione peroxidase 4 (GPX4), leading to membrane rupture and cell death [119].
Iron can catalyze ferroptosis both enzymatically and non-enzymatically. Enzymatic catalysis involves facilitating the oxidation of lipids within cells, forming lipid peroxides [120]. Autophagic degradation of ferritin (an iron-storage protein) results in the leakage of free iron (Fe2+) into the cytoplasm, thereby expanding the pool of redox-active iron and enhancing susceptibility to ferroptosis. The overexpression of transferrin receptor 1 (TfR1) enhances the amount of iron that is internalized and enhances susceptibility to ferroptosis [121]. The main factors involved in the process of lipid peroxidation include lipoxygenases and phosphorylase kinase G2 (PHKG2), which regulate the availability of iron required to catalyze the peroxidation of polyunsaturated fatty acids (PUFAs) [122].
In addition to GPX4, the cystine-glutamate antiporter SLC7A11 and ferroptosis suppressor protein 1 (FSP1) contribute significantly to the inhibition of ferroptosis via lipid peroxide detoxification [123]. Both genetic alterations and chemical inhibition of antioxidant defenses trigger the induction of ferroptosis [124]. The main sources of ROS generation during ferroptosis are activated mitochondria, NADPH oxidases, and the Fenton reaction [125]. Lipid peroxidation increases the levels of lipid ROS, forming a vicious cycle that increases cell death [126]. Mitochondria are a major source of ROS, and disruption of iron metabolism can trigger ferroptosis [125]. Inhibition of mitochondrial oxidative phosphorylation further increases intracellular ROS levels [120,127,128,129]. A flow model of the steps in ferroptosis involving various membrane and intracellular components including transferrin receptors, glutamine-cystine antiporter (Xc), LOX, GPX, GSH, iron, and lipid peroxides, is presented (Figure 1). A summary of the various molecular mechanisms of ferroptosis is presented, including details such as genes and pathways (Table 1 and Figure 2).

3. Types of Ferroptosis

Ferroptosis is traditionally considered a uniform cell death process involving multiple regulatory pathways [127,128]. However, distinct mechanistic variations exist, reflecting how cells manage oxidative stress, iron overload, or lipid peroxidation, suggesting pathway-dependent or context-specific subtypes [129,130].
Classical ferroptosis is the most thoroughly studied form, in which ferroptosis is initiated through lipid peroxidation and iron-catalyzed reactions and failure of the antioxidant defense system, primarily glutathione peroxidase 4 (GPX4). The major contributors are iron overload and glutathione depletion, with the accumulation of lipid peroxides [124,125]. Under conditions of GSH depletion, such as during oxidative stress or inhibition of the cystine-glutamate antiporter (system Xc−), GPX4 is also inhibited. This results in the accumulation of lipid peroxides and eventual ferroptotic cell death [129]. Iron plays a central role in catalyzing Fenton reactions to produce hydroxyl radicals that intensify lipid peroxidation [131]. Whereas glutathione plays a critical role as a cofactor for GPX4, an antioxidant that neutralizes ROS and lipid ROS [132].
Mitochondria-dependent ferroptosis is implicated in the promotion of oxidative damage due to mitochondria dysfunction [133]. Mitochondria are the power source for cells, which are not only essential for energy production but also for regulating respiration and the production of mitochondria-specific ROS, which accordingly heightens susceptibility to ferroptosis [134]. This pathway can be triggered by the disruption of mitochondria function due to various stressors, iron accumulation, or specific inhibitors of gamma-glutamylcysteine synthase, such as buthionine sulfoximine (BSO), which depletes GSH, destroys mitochondrial functions, and elevates ROS levels. This ultimately leads to an increase in ROS and oxidative damage to cellular lipids, inducing ferroptosis through a mitochondria-dependent pathway.
FSP1, also known as ferroptosis suppressor protein1, has been described as a mechanism of protecting cells from ferroptosis, independent of the activities of GPX4 [135]. It contains CoQ10 (ubiquinone), a lipid-soluble antioxidant that prevents lipid peroxidation, recycles CoQ10, thereby regenerating its antioxidant capacity and providing an additional defense mechanism. Cells lacking functioning GPX4 may undergo ferroptosis through the FSP1 pathway [136].
NADPH oxidase (NOX)-dependent ferroptosis is associated with NOX catalytic activity, leading to the generation of ROS by transferring electrons from NADPH to oxygen, producing superoxide and ROS, which damage cellular membranes [137]. NADPH oxidase enzymes produce ROS, leading to lipid peroxidation and ferroptosis [138]. There is a link between NOX activity and the induction of ferroptosis, especially via the mediation of the YAP-TAZ signaling pathway [139]. NOX inhibitors are suggested to inhibit ferroptosis, especially in cells with high NOX activity. The enhancement of NOX activity can lead to an increase in ferroptosis [140]. NOX-dependent ferroptosis may function as a distinct regulatory mechanism, offering potential therapeutic applications in diseases with disrupted NOX activity.
Iron overload-induced ferroptosis results from unregulated oxidative damage via the Fenton reaction, in which iron catalyzes hydroxyl radical formation, triggering lipid peroxidation. This occurs in cells lacking proper iron storage mechanisms, such as ferritin levels [141]. This kind of ferroptosis is due to imbalanced iron homeostasis, for example, through ferritin degradation, induction of the transferrin receptor, or accumulation of excessive iron [142]. Chelators of iron, such as deferoxamine, also repress it by reducing free iron concentrations and preventing the Fenton reaction from occurring [143].
Erastin-induced ferroptosis is linked to the blockade of system Xc−, an antiporter of cystine/glutamate that plays a major role in maintaining cellular glutathione levels [144]. Erastin disrupts the synthesis of glutathione by inhibiting cysteine uptake, leading to the depletion of this antioxidant [145]. Glutathione serves as a cofactor for GPX4 and is required for its activity of GPX4 [146]. This pathway is important because it links the disruption of cellular antioxidant defenses with ferroptosis initiation. It has major applications in cancer treatment because it can enhance the efficacy of chemotherapy and other treatments.
Radiation-induced ferroptosis has recently been indicated to cause ferroptosis via exposure to ionizing radiation, leading to the generation of ROS, particularly lipid peroxidation of iron-loaded cells [147]. This mechanism remains of significant interest in cancer therapy, given that the induction of ferroptosis renders radiation therapy more potent [148]. Irradiation prompts iron-dependent generation of ROS and subsequent lipid peroxidation [149]. A summary of the various types of ferroptosis is provided, including descriptions of key genes and cancer hallmarks (Table 2 and Figure 3).

3.1. Ferroptosis: Detection, Estimation, and Biomarkers of Ferroptosis

The detection and estimation of ferroptosis assume major importance in understanding its role in such diseases and devising therapeutic strategies [150,151,152,153,154,155,156]. A particular fluorescent probe, PPAC-C4, has been developed for the dual ratio and ultrahigh-accuracy quantification of mitochondrial viscosity, which is enhanced during ferroptosis [150]. To track the reversal of polarity in the plasma membrane during ferroptosis, polarized fluorescent probes Mem-C1C18 and Mem-C18C18 were developed. In high-resolution fluorescence labeling and quantification, Mem-C1C18 was shown to be superior to C18C18 [151].
Detecting ferroptosis in cells requires specific biomarkers that are unique to lipid peroxidation and iron metabolism [152]. Common methods of detection include measuring lipid peroxidation products, such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), and using iron-sensitive dyes to detect intracellular iron [153]. Measuring these markers can help detect cells in the process of ferroptosis [154]. The cystine/glutamate antiporter SLC7A11 (also commonly known as xCT) is reported to mediate metabolic reprogramming in cancer by impinging on ferroptosis-dependent cell death pathways [155].
In recent years, the identification of biomarkers of ferroptosis has attracted great attention, as they can significantly enhance chemotherapy by providing tools to monitor, predict, and potentially mitigate cell death related to iron-dependent lipid peroxidation. Several oxidative stress biomarkers have now been identified that appear to be present during ferroptosis. For example, ophthalamic acid has been detected in blood following depletion of GSH in the liver and is now considered a biomarker of oxidative stress [156]. We have also detected the presence of opthalamates during the process of ferroptosis in ovarian cancer cells [157]. In addition, modulation of carnitine levels has been observed during ferroptosis in both colon and ovarian cells [158]. Various genes are induced during oxidative stress that are also induced during ferroptosis. Of particular interest is the CHAC1 gene, a master regulator of oxidative stress [159], which is significantly induced during ferroptosis in colon [158] and ovarian cancer cells [160].
Advancements in fluorescent probes and multiplexed assays have significantly improved ferroptosis detection [161,162]. Precise monitoring of molecular targets enhances the understanding of their role in disease and aids in the development of targeted therapy [163]. Organic fluorescent probes can detect multiple biomolecules and microenvironments during ferroptosis, offering non-destructive and easy-to-prepare tools for assessing homeostasis and physiological changes [162,163].

3.2. Ferroptosis: Preclinical Evidence

Preclinical and clinical researchers have generated significant interest in modulating ferroptosis in diseases such as cancer as a new avenue of therapy [81,82,83,84,85,86,87,88,89,90]. Combining chemotherapy with ferroptosis inducers is believed to be an improved strategy to overcome chemotherapy resistance and enhance therapeutic efficacy [164,165,166]. It is now recognized as a promising avenue for cancer treatment in the clinic [167,168,169,170,171,172,173,174,175,176]. PRLX93936, an analog of erastin, has been tested in clinical trials. Co-treatment with cisplatin and PRLX93936 induces lipid peroxidation and Fe2+ production, thereby promoting ferroptosis [165].
Ferroptosis inducers, such as erastin and its analogs, have shown promise in sensitizing resistant tumor cells to various chemotherapeutic agents [166,167,168,169,170,171,172]. Our recent work has shown that erastin, as a single agent, is highly effective against both P-gp- and BCRP-expressing cell lines [164,167]. However, RSL3, an inhibitor of GPX4, was significantly more cytotoxic to these cells [167]. Furthermore, both erastin and RSL3 significantly enhanced the cytotoxicity of adriamycin and topotecan in these cell lines [167]. Yang et al. reported that CHAC1 (glutathione-specific γ-glutamylcyclotransferase 1) induction significantly enhanced radiation-dependent ferroptosis in thyroid cancer cells [168]. Recently, ML162 and ML210 have been indicated to lack an inhibitory effect on selenoprotein GPX4. However, ML162 and ML210 were found to be efficient inhibitors of another selenoprotein, TXNRD1 [169].
Inducers of ferroptosis are also implicated in the modulation of tumor responses, including immune responses [170]. Mitigation of GPX4 by lentivirus sh-GPX4 in TNBC cells enhances sensitivity to gefitinib by modulating ferroptosis [171]. In cisplatin-resistant ovarian cancer cells, tripterygium glycosides disturb redox homeostasis and enhance ferroptosis, leading to increased chemosensitivity [172]. In ovarian cancer cells, the combinatorial effects of NRF2 inhibitors and GPX4 inhibitors resulted in the suppression of growth and decreased formation of spheroids over anticancer drugs alone in cell culture and 3D model, respectively [173]. In HCC cells, silencing of soluble vector family member 6 (SLC2A6) leads to the suppression of proliferation, migration, and invasion, and these observations are linked to the ferroptosis pathway [174]. Additionally, the combination of asiatic acid and sorafenib enhances ferroptosis in HCC cells by mediating JNK1/2 signaling [175].
A summary of data on the various inducers of ferroptosis in combination with anticancer drugs is presented (Table 3).

3.3. Ferroptosis: Clinical Evidence

Ferroptosis has emerged as a promising strategy for cancer treatment in clinical settings [167,168,169,170,171,172,173]. However, most clinical applications remain in the investigation stage. Among the most encouraging directions are preclinical studies showing synergistic effects between ferroptosis inducers and existing chemotherapeutic agents, highlighting the potential of combination therapy [165,166]. While robust preclinical data support ferroptosis as a viable anticancer approach, its translation into clinical practice is still in the early stages, albeit gaining momentum. Clinical evidence of the induction of ferroptosis in human diseases such as epilepsy, mucositis, and autoimmune diseases has been recorded in the Clinical Trial Registry. However, clinical data on the induction of ferroptosis, either as monotherapy or in combination with therapies, are limited. For example, sulfasalazine, a system Xc− inhibitor, is currently under investigation in trials targeting solid tumors and glioblastoma (NCT04205357) [177]. Another clinical interventional study is in progress on the use of carbon nanoparticle-loaded iron [CNSI-Fe(II)], which can induce ferroptosis in patients with advanced solid tumors (NCT06048367) [178].

4. Conclusions and Future Perspectives

Ferroptosis is a promising cancer therapy strategy that offers a means to overcome treatment resistance and selectively target cancer cells. Chemotherapy combined with ferroptosis inducers enhances therapeutic efficacy, particularly in drug-resistant cancers like ovarian, pancreatic, and colorectal malignancies. These cancers exhibit altered iron metabolism and oxidative stress, making them more susceptible to ferroptosis. Ferroptosis inducers, such as erastin (xCT inhibitor) and RSL3 (GPX4 inhibitor), work synergistically with chemotherapy by increasing lipid peroxidation and intracellular iron levels. Additionally, ferroptosis enhances immune responses by exposing tumor antigens and improving immunotherapy outcomes.
Despite its potential, there are still challenges to be addressed. For example, erastin has poor water solubility and undergoes rapid metabolism, which limits its clinical success. Further research is needed to optimize delivery systems and explore the epigenetic regulation of ferroptosis pathways.
Future studies should investigate new therapeutic approaches, including biomimetic-engineered bacterial conjugates and magnetotactic bacteria as targeted ferroptosis inducers.
Identifying ferroptosis biomarkers is crucial for personalized medicine, enabling tailored treatments that combine ferroptosis inducers with chemotherapy to achieve maximum efficacy and minimal side effects.
AI and machine learning can accelerate drug discovery and combinatorial therapy design, helping overcome multidrug resistance. Integrating ferroptosis into cancer treatment could revolutionize precision oncology and improve patient outcomes.
In the future, better-engineered nano-carriers and nanodrug delivery systems could be employed to improve the effectiveness of ferroptosis inducers.

Author Contributions

M.S., Drafting and Writing; H.L.A., Drafting and Writing; R.N., Drafting and Writing; S.J., Drafting and Writing; K.S., Drafting and Writing; N.K.S., Conceptualization, Writing, and Editing; and B.K.S., Conceptualization, Writing, and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the intramural research program (Grant number ZIA E505013922 and CARCI-HEI#) of the National Institute of Environmental Health Sciences, NIH. The statements contained herein do not necessarily represent the statements, opinions, or conclusions of the NIEHS, NIH, or the US government.

Data Availability Statement

The authors declare that no data generated and submitted and hence not applicable.

Acknowledgments

The authors thank Erik Tokar and Brian Silver for their critical evaluation of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dagogo-Jack, I.; Shaw, A.T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 2018, 15, 81–94. [Google Scholar] [CrossRef] [PubMed]
  2. Ge, R.; Wang, Z.; Cheng, L. Tumor microenvironment heterogeneity: An important mediator of prostate cancer progression and therapeutic resistance. NPJ Precis. Oncol. 2022, 6, 31. [Google Scholar] [CrossRef] [PubMed]
  3. Khan, S.U.; Fatima, K.; Aisha, S.; Malik, F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun. Signal. 2024, 22, 109. [Google Scholar] [CrossRef]
  4. Liu, S.; Zhang, X.; Wang, W.; Li, X.; Sun, X.; Zhao, Y.; Wang, Q.; Li, Y.; Hu, F.; Ren, H. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol. Cancer 2024, 23, 261. [Google Scholar]
  5. Yang, S.; Hu, C.; Chen, X.; Tang, Y.; Li, J.; Yang, H.; Yang, Y.; Ying, B.; Xiao, X.; Li, S.Z.; et al. Crosstalk between metabolism and cell death in tumorigenesis. Mol. Cancer 2024, 23, 71. [Google Scholar] [CrossRef] [PubMed]
  6. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
  7. Chen, Z.; Wang, W.; Abdul Razak, S.R.; Han, T.; Ahmad, N.H.; Li, X. Ferroptosis as a potential target for cancer therapy. Cell Death Dis. 2023, 14, 460. [Google Scholar] [CrossRef] [PubMed]
  8. Parlani, M.; Jorgez, C.; Friedl, P. Plasticity of cancer invasion and energy metabolism. Trends Cell Biol. 2023, 33, 388–402. [Google Scholar] [CrossRef]
  9. Mao, C.; Wang, M.; Zhuang, L.; Gan, B. Metabolic cell death in cancer: Ferroptosis, cuproptosis, disulfidptosis, and beyond. Protein Cell 2024, 15, 642–660. [Google Scholar] [CrossRef] [PubMed]
  10. NaveenKumar, S.K.; SharathBabu, B.N.; Hemshekhar, M.; Kemparaju, K.; Girish, K.S.; Mugesh, G. The role of reactive oxygen species and ferroptosis in heme-mediated activation of human platelets. ACS Chem. Biol. 2018, 13, 1996–2002. [Google Scholar] [CrossRef]
  11. Lyamzaev, K.G.; Panteleeva, A.A.; Simonyan, R.A.; Avetisyan, A.V.; Chernyak, B.V. Mitochondrial lipid peroxidation is responsible for ferroptosis. Cells 2023, 12, 611. [Google Scholar] [CrossRef]
  12. Beloribi-Djefaflia, S.; Vasseur, S.; Guillaumond, F. Lipid metabolic reprogramming in cancer cells. Oncogenesis 2016, 5, e189. [Google Scholar] [CrossRef] [PubMed]
  13. Bertrand, R.L. Iron accumulation, glutathione depletion, and lipid peroxidation must occur simultaneously during ferroptosis and are mutually amplifying events. Med. Hypotheses 2017, 101, 69–74. [Google Scholar] [CrossRef] [PubMed]
  14. Li, D.; Li, Y. The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct. Target. Ther. 2020, 5, 108. [Google Scholar]
  15. Morales, M.; Xue, X. Targeting Iron Metabolism in Cancer Therapy. Theranostics 2021, 11, 8412–8429. [Google Scholar] [CrossRef]
  16. Tong, X.; Tang, R.; Xiao, M.; Xu, J.; Wang, W.; Zhang, B.; Liu, J.; Yu, X.; Shi, S. Targeting Cell Death Pathways for Cancer Therapy: Recent Developments in Necroptosis, Pyroptosis, Ferroptosis, and Cuproptosis Research. J. Hematol. Oncol. 2022, 15, 174. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, C.; Liu, X.; Jin, S.; Chen, Y.; Guo, R. Ferroptosis in Cancer Therapy: A Novel Approach to Reversing Drug Resistance. Mol. Cancer 2022, 21, 47. [Google Scholar] [CrossRef]
  18. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  19. Fouad, Y.A.; Aanei, C. Revisiting the Hallmarks of Cancer. Am. J. Cancer Res. 2017, 7, 1016–1036. [Google Scholar]
  20. Pranzini, E.; Pardella, E.; Paoli, P.; Fendt, S.M.; Taddei, M.L. Metabolic Reprogramming in Anticancer Drug Resistance: A Focus on Amino Acids. Trends Cancer 2021, 7, 682–699. [Google Scholar] [CrossRef]
  21. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  22. Stine, Z.E.; Schug, Z.T.; Salvino, J.M.; Dang, C.V. Targeting Cancer Metabolism in the Era of Precision Oncology. Nat. Rev. Drug Discov. 2022, 21, 141–162. [Google Scholar] [CrossRef] [PubMed]
  23. El-Tanani, M.; Rabbani, S.A.; El-Tanani, Y.; Matalka, I.I. Metabolic Vulnerabilities in Cancer: A New Therapeutic Strategy. Crit. Rev. Oncol. Hematol. 2024, 201, 104438. [Google Scholar] [CrossRef]
  24. Mortensen, M.S.; Ruiz, J.; Watts, J.L. Polyunsaturated Fatty Acids Drive Lipid Peroxidation during Ferroptosis. Cells 2023, 12, 804. [Google Scholar] [CrossRef]
  25. Sun, S.; Shen, J.; Jiang, J.; Wang, F.; Min, J. Targeting Ferroptosis Opens New Avenues for the Development of Novel Therapeutics. Signal Transduct. Target. Ther. 2023, 8, 372. [Google Scholar] [CrossRef]
  26. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
  27. Chen, Y.; Fan, Z.; Yang, Y.; Gu, C. Iron Metabolism and Its Contribution to Cancer. Int. J. Oncol. 2019, 54, 1143–1154. [Google Scholar] [CrossRef] [PubMed]
  28. Zheng, J.; Conrad, M. The Metabolic Underpinnings of Ferroptosis. Cell Metab. 2020, 32, 920–937. [Google Scholar] [CrossRef]
  29. Kazan, H.H.; Urfali-Mamatoglu, C.; Gunduz, U. Iron Metabolism and Drug Resistance in Cancer. Biometals 2017, 30, 629–641. [Google Scholar] [CrossRef]
  30. Kawahara, I.; Yoshino, H.; Fukumoto, W.; Arima, J.; Saito, S.; Li, G.; Fukuda, I.; Mitsuke, A.; Sakaguchi, T.; Inoguchi, S.; et al. Targeting Metabolic Reprogramming to Overcome Drug Resistance in Advanced Bladder Cancer: Insights from Gemcitabine- and Cisplatin-Resistant Models. Mol. Oncol. 2024, 18, 2196–2211. [Google Scholar] [CrossRef]
  31. Burz, C.; Berindan-Neagoe, I.; Balacescu, O.; Irimie, A. Apoptosis in Cancer: Key Molecular Signaling Pathways and Therapy Targets. Acta Oncol. 2009, 48, 811–821. [Google Scholar] [CrossRef] [PubMed]
  32. Brown, J.M.; Wilson, G. Apoptosis Genes and Resistance to Cancer Therapy: What Does the Experimental and Clinical Data Tell Us? Cancer Biol. Ther. 2003, 2, 477–490. [Google Scholar] [CrossRef]
  33. Bhat, G.R.; Sethi, I.; Sadida, H.Q.; Rah, B.; Mir, R.; Algehainy, N.; Albalawi, I.A.; Masoodi, T.; Subbaraj, G.K.; Jamal, F.; et al. Cancer Cell Plasticity: From Cellular, Molecular, and Genetic Mechanisms to Tumor Heterogeneity and Drug Resistance. Cancer Metastasis Rev. 2024, 43, 197–228. [Google Scholar] [CrossRef]
  34. Jin, Z.; El-Deiry, W.S. Overview of Cell Death Signaling Pathways. Cancer Biol. Ther. 2005, 4, 139–163. [Google Scholar] [CrossRef] [PubMed]
  35. Maddika, S.; Ande, S.R.; Panigrahi, S.; Paranjothy, T.; Weglarczyk, K.; Zuse, A.; Eshraghi, M.; Manda, K.D.; Wiechec, E.; Los, M. Cell Survival, Cell Death and Cell Cycle Pathways Are Interconnected: Implications for Cancer Therapy. Drug Resist. Updates 2007, 10, 13–29. [Google Scholar] [CrossRef]
  36. Liu, Z.G.; Jiao, D. Necroptosis, Tumor Necrosis and Tumorigenesis. Cell Stress 2019, 4, 1–8. [Google Scholar] [CrossRef] [PubMed]
  37. Schmitt, C.A.; Wang, B.; Demaria, M. Senescence and Cancer—Role and Therapeutic Opportunities. Nat. Rev. Clin. Oncol. 2022, 19, 619–636. [Google Scholar] [CrossRef]
  38. Debnath, J.; Gammoh, N.; Ryan, K.M. Autophagy and Autophagy-Related Pathways in Cancer. Nat. Rev. Mol. Cell Biol. 2023, 24, 560–575. [Google Scholar] [CrossRef]
  39. McGee, M.M. Targeting the Mitotic Catastrophe Signaling Pathway in Cancer. Mediat. Inflamm. 2015, 2015, 146282. [Google Scholar] [CrossRef]
  40. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular Mechanisms of Cell Death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar]
  41. Wang, X.; Hua, P.; He, C.; Chen, M. Non-apoptotic cell death-based cancer therapy: Molecular mechanism, pharmacological modulators, and nanomedicine. Acta Pharm. Sin. B 2022, 12, 3567–3593. [Google Scholar] [CrossRef] [PubMed]
  42. Fulda, S. Tumor resistance to apoptosis. Int. J. Cancer 2009, 124, 511–515. [Google Scholar] [CrossRef] [PubMed]
  43. Kar, A.; Agarwal, S.; Singh, A.; Bajaj, A.; Dasgupta, U. Insights into molecular mechanisms of chemotherapy resistance in cancer. Transl. Oncol. 2024, 42, 101901. [Google Scholar] [CrossRef]
  44. McIlwain, D.R.; Berger, T.; Mak, T.W. Caspase functions in cell death and disease. Cold Spring Harb. Perspect. Biol. 2013, 5, a008656. [Google Scholar] [CrossRef]
  45. Ozaki, T.; Nakagawara, A. Role of p53 in cell death and human cancers. Cancers 2011, 3, 994–1013. [Google Scholar] [CrossRef]
  46. Qian, S.; Wei, Z.; Yang, W.; Huang, J.; Yang, Y.; Wang, J. The role of BCL-2 family proteins in regulating apoptosis and cancer therapy. Front. Oncol. 2022, 12, 985363. [Google Scholar] [CrossRef] [PubMed]
  47. Sazonova, E.V.; Yapryntseva, M.A.; Pervushin, N.V.; Tsvetcov, R.I.; Zhivotovsky, B.; Kopeina, G.S. Cancer Drug Resistance: Targeting Proliferation or Programmed Cell Death. Cells 2024, 13, 388. [Google Scholar] [CrossRef]
  48. Kerr, J.F.; Winterford, C.M.; Harmon, B.V. Apoptosis: Its significance in cancer and cancer therapy. Cancer 1994, 73, 2013–2026. [Google Scholar] [CrossRef]
  49. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef]
  50. Levine, A.J.; Oren, M. The first 30 years of p53: Growing ever more complex. Nat. Rev. Cancer 2009, 9, 749–758. [Google Scholar] [CrossRef]
  51. Negrini, S.; Gorgoulis, V.G.; Halazonetis, T.D. Genomic instability—An evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 2010, 11, 220–228. [Google Scholar] [CrossRef] [PubMed]
  52. Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef]
  53. Zhu, C.; Fan, F.; Li, C.Y.; Xiong, Y.; Liu, X. Caspase-3 promotes oncogene-induced malignant transformation via EndoG-dependent Src-STAT3 phosphorylation. Cell Death Dis. 2024, 15, 486. [Google Scholar] [CrossRef] [PubMed]
  54. Fuchs, Y.; Steller, H. Programmed cell death in animal development and disease. Cell 2011, 147, 742–758. [Google Scholar] [CrossRef] [PubMed]
  55. Kashyap, D.; Garg, V.K.; Goel, N. Intrinsic and extrinsic pathways of apoptosis: Role in cancer development and prognosis. Adv. Protein Chem. Struct. Biol. 2021, 125, 73–120. [Google Scholar]
  56. Zhang, J.; Lou, X.; Jin, L.; Zhou, R.; Liu, S.; Xu, N.; Liao, D.J. Necrosis, and then stress induced necrosis-like cell death, but not apoptosis, should be the preferred cell death mode for chemotherapy: Clearance of a few misconceptions. Oncoscience 2014, 1, 407–422. [Google Scholar] [CrossRef]
  57. Ketelut-Carneiro, N.; Fitzgerald, K.A. Apoptosis, Pyroptosis, and Necroptosis—Oh My! The Many Ways a Cell Can Die. J. Mol. Biol. 2022, 434, 167378. [Google Scholar] [CrossRef]
  58. Shahar, N.; Larisch, S. Inhibiting the inhibitors: Targeting anti-apoptotic proteins in cancer and therapy resistance. Drug Resist. Updates 2020, 52, 100712. [Google Scholar] [CrossRef]
  59. Mantovani, F.; Collavin, L.; Del Sal, G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ. 2019, 26, 199–212. [Google Scholar] [CrossRef]
  60. Carneiro, B.A.; El-Deiry, W.S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 2020, 17, 395–417. [Google Scholar] [CrossRef]
  61. Ryter, S.W.; Cloonan, S.M.; Choi, A.M. Autophagy: A critical regulator of cellular metabolism and homeostasis. Mol. Cells 2013, 36, 7–16. [Google Scholar] [CrossRef] [PubMed]
  62. Chun, Y.; Kim, J. Autophagy: An Essential Degradation Program for Cellular Homeostasis and Life. Cells 2018, 7, 278. [Google Scholar] [CrossRef]
  63. Chang, H.; Zou, Z. Targeting autophagy to overcome drug resistance: Further developments. J. Hematol. Oncol. 2020, 13, 159. [Google Scholar] [CrossRef] [PubMed]
  64. Amaravadi, R.K.; Kimmelman, A.C.; Debnath, J. Targeting Autophagy in Cancer: Recent Advances and Future Directions. Cancer Discov. 2019, 9, 1167–1181. [Google Scholar] [CrossRef] [PubMed]
  65. Paquette, M.; El-Houjeiri, L.; Pause, A. mTOR Pathways in Cancer and Autophagy. Cancers 2018, 10, 18. [Google Scholar] [CrossRef]
  66. Bhutia, S.K.; Mukhopadhyay, S.; Sinha, N.; Das, D.N.; Panda, P.K.; Patra, S.K.; Maiti, T.K.; Mandal, M.; Dent, P.; Wang, X.Y.; et al. Autophagy: Cancer’s Friend or Foe? Adv. Cancer Res. 2013, 118, 61–95. [Google Scholar]
  67. Xu, Z.; Han, X.; Ou, D.; Liu, T.; Li, Z.; Jiang, G.; Liu, J.; Zhang, J. Targeting PI3K/AKT/mTOR-mediated autophagy for tumor therapy. Appl. Microbiol. Biotechnol. 2020, 104, 575–587. [Google Scholar] [CrossRef]
  68. Altman, B.J.; Rathmell, J.C. Metabolic stress in autophagy and cell death pathways. Cold Spring Harb. Perspect. Biol. 2012, 4, a008763. [Google Scholar] [CrossRef]
  69. Zhang, J.; Chen, A.; Song, Y. Propofol Triggers Cell Death in Lung Cancer Cells by Increasing PANX1 Expression, Activating the Mitochondrial Cell Death Pathway, and Enhancing ROS Levels. Disc Med. 2024, 36, 2231–2243. [Google Scholar] [CrossRef]
  70. Shen, S.; Shao, Y.; Li, C. Different types of cell death and their shift in shaping disease. Cell Death Discov. 2023, 9, 284. [Google Scholar] [CrossRef]
  71. D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef]
  72. Fulda, S.; Gorman, A.M.; Hori, O.; Samali, A. Cellular stress responses: Cell survival and cell death. Int. J. Cell Biol. 2010, 2010, 214074. [Google Scholar] [CrossRef]
  73. Brault, M.; Olsen, T.M.; Martinez, J.; Stetson, D.B.; Oberst, A. Intracellular Nucleic Acid Sensing Triggers Necroptosis through Synergistic Type I IFN and TNF Signaling. J. Immunol. 2018, 200, 2748–2756. [Google Scholar] [CrossRef] [PubMed]
  74. Shin, H.J.; Kwon, H.K.; Lee, J.H.; Gui, X.; Achek, A.; Kim, J.H.; Choi, S. Doxorubicin-induced necrosis is mediated by poly-(ADP-ribose) polymerase 1 (PARP1) but is independent of p53. Sci. Rep. 2015, 5, 15798. [Google Scholar] [CrossRef]
  75. Garay, R.P.; Viens, P.; Bauer, J.; Normier, G.; Bardou, M.; Jeannin, J.F.; Chiavaroli, C. Cancer relapse under chemotherapy: Why TLR2/4 receptor agonists can help. Eur. J. Pharmacol. 2007, 563, 1–17. [Google Scholar] [CrossRef] [PubMed]
  76. Ou, L.; Lin, S.; Song, B.; Liu, J.; Lai, R.; Shao, L. The Mechanisms of Graphene-Based Materials-Induced Programmed Cell Death: A Review of Apoptosis, Autophagy, and Programmed Necrosis. Int. J. Nanomed. 2017, 12, 6633–6646. [Google Scholar] [CrossRef] [PubMed]
  77. Ploumi, C.; Papandreou, M.E.; Tavernarakis, N. The Complex Interplay between Autophagy and Cell Death Pathways. Biochem. J. 2022, 479, 75–90. [Google Scholar] [CrossRef]
  78. Christofferson, D.E.; Yuan, J. Necroptosis as an Alternative Form of Programmed Cell Death. Curr. Opin. Cell Biol. 2010, 22, 263–268. [Google Scholar] [CrossRef]
  79. Bansal, N.; Sciabola, S.; Bhisetti, G. Understanding Allosteric Interactions in hMLKL Protein That Modulate Necroptosis and Its Inhibition. Sci. Rep. 2019, 9, 16853. [Google Scholar] [CrossRef]
  80. Newton, K.; Strasser, A.; Kayagaki, N.; Dixit, V.M. Cell Death. Cell 2024, 187, 235–256. [Google Scholar] [CrossRef]
  81. Nikoletopoulou, V.; Markaki, M.; Palikaras, K.; Tavernarakis, N. Crosstalk between Apoptosis, Necrosis and Autophagy. Biochim. Biophys. Acta 2013, 1833, 3448–3459. [Google Scholar] [CrossRef] [PubMed]
  82. Pu, F.; Chen, F.; Zhang, Z.; Shi, D.; Zhong, B.; Lv, X.; Tucker, A.B.; Fan, J.; Li, A.J.; Qin, K.; et al. Ferroptosis as a Novel Form of Regulated Cell Death: Implications in the Pathogenesis, Oncometabolism and Treatment of Human Cancer. Genes Dis. 2020, 9, 347–357. [Google Scholar] [CrossRef]
  83. Yan, H.F.; Zou, T.; Tuo, Q.Z.; Xu, S.; Li, H.; Belaidi, A.A.; Lei, P. Ferroptosis: Mechanisms and Links with Diseases. Signal Transduct. Target. Ther. 2021, 6, 49. [Google Scholar] [CrossRef] [PubMed]
  84. Jin, X.; Tang, J.; Qiu, X.; Nie, X.; Ou, S.; Wu, G.; Zhang, R.; Zhu, J. Ferroptosis: Emerging Mechanisms, Biological Function, and Therapeutic Potential in Cancer and Inflammation. Cell Death Discov. 2024, 10, 45. [Google Scholar] [CrossRef]
  85. Chen, Y.; Guo, X.; Zeng, Y.; Mo, X.; Hong, S.; He, H.; Li, J.; Fatima, S.; Liu, Q. Oxidative Stress Induces Mitochondrial Iron Overload and Ferroptotic Cell Death. Sci. Rep. 2023, 13, 15515. [Google Scholar] [CrossRef] [PubMed]
  86. Krishnamurthy, H.K.; Pereira, M.; Rajavelu, I.; Jayaraman, V.; Krishna, K.; Wang, T.; Bei, K.; Rajasekaran, J.J. Oxidative Stress: Fundamentals and Advances in Quantification Techniques. Front. Chem. 2024, 12, 1470458. [Google Scholar] [CrossRef]
  87. Jena, A.B.; Samal, R.R.; Bhol, N.K.; Duttaroy, A.K. Cellular Red-Ox System in Health and Disease: The Latest Update. Biomed. Pharmacother. 2023, 162, 114606. [Google Scholar] [CrossRef]
  88. Liu, Y.; Zhou, L.; Xu, Y.; Li, K.; Zhao, Y.; Qiao, H.; Xu, Q.; Zhao, J. Heat Shock Proteins and Ferroptosis. Front. Cell Dev. Biol. 2022, 10, 864635. [Google Scholar] [CrossRef]
  89. Lei, G.; Zhang, Y.; Koppula, P.; Liu, X.; Zhang, J.; Lin, S.H.; Ajani, J.A.; Xiao, Q.; Liao, Z.; Wang, H.; et al. The Role of Ferroptosis in Ionizing Radiation-Induced Cell Death and Tumor Suppression. Cell Res. 2020, 30, 146–162. [Google Scholar] [CrossRef]
  90. Mbah, N.E.; Lyssiotis, C.A. Metabolic Regulation of Ferroptosis in the Tumor Microenvironment. J. Biol. Chem. 2022, 298, 101617. [Google Scholar] [CrossRef]
  91. Vana, F.; Szabo, Z.; Masarik, M.; Kratochvilova, M. The Interplay of Transition Metals in Ferroptosis and Pyroptosis. Cell Div. 2024, 19, 24. [Google Scholar] [CrossRef]
  92. Qi, D.; Peng, M. Ferroptosis-Mediated Immune Responses in Cancer. Front. Immunol. 2023, 14, 1188365. [Google Scholar] [CrossRef]
  93. Vucetic, M.; Daher, B.; Cassim, S.; Meira, W.; Pouyssegur, J. Together We Stand, Apart We Fall: How Cell-to-Cell Contact/Interplay Provides Resistance to Ferroptosis. Cell Death Dis. 2020, 11, 789. [Google Scholar] [CrossRef] [PubMed]
  94. Bano, I.; Horky, P.; Abbas, S.Q.; Majid, M.; Bilal, A.H.M.; Ali, F.; Behl, T.; Hassan, S.S.U.; Bungau, S. Ferroptosis: A New Road towards Cancer Management. Molecules 2022, 27, 2129. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, Z.; Lin, H.; Wang, X.; Li, G.; Liu, N.; Zhang, M.; Shen, Y. The Application of Approaches in Detecting Ferroptosis. Heliyon 2023, 10, e23507. [Google Scholar] [CrossRef] [PubMed]
  96. Veglia Tranchese, R.; Battista, S.; Cerchia, L.; Fedele, M. Ferroptosis in Cancer: Epigenetic Control and Therapeutic Opportunities. Biomolecules 2024, 14, 1443. [Google Scholar] [CrossRef]
  97. Bebber, C.M.; Müller, F.; Prieto Clemente, L.; Weber, J.; von Karstedt, S. Ferroptosis in Cancer Cell Biology. Cancers 2020, 12, 164. [Google Scholar] [CrossRef]
  98. Gooz, M.; Maldonado, E.N. Fluorescence Microscopy Imaging of Mitochondrial Metabolism in Cancer Cells. Front. Oncol. 2023, 13, 1152553. [Google Scholar] [CrossRef]
  99. Bianchi, M.E.; Manfredi, A. Chromatin and Cell Death. Biochim. Biophys. Acta 2004, 1677, 181–186. [Google Scholar] [CrossRef]
  100. Sharma, A.; Flora, S.J.S. Positive and Negative Regulation of Ferroptosis and Its Role in Maintaining Metabolic and Redox Homeostasis. Oxid. Med. Cell. Longev. 2021, 2021, 9074206. [Google Scholar] [CrossRef]
  101. Kroemer, G.; Martin, S.J. Caspase-Independent Cell Death. Nat. Med. 2005, 11, 725–730. [Google Scholar] [CrossRef] [PubMed]
  102. Cai, Z.; Liu, Z.G. Execution of RIPK3-Regulated Necrosis. Mol. Cell. Oncol. 2014, 1, e960759. [Google Scholar] [CrossRef] [PubMed]
  103. Gao, W.; Wang, X.; Zhou, Y.; Wang, X.; Yu, Y. Autophagy, Ferroptosis, Pyroptosis, and Necroptosis in Tumor Immunotherapy. Signal Transduct. Target. Ther. 2022, 7, 196. [Google Scholar] [CrossRef] [PubMed]
  104. Lei, G.; Zhuang, L.; Gan, B. The Roles of Ferroptosis in Cancer: Tumor Suppression, Tumor Microenvironment, and Therapeutic Interventions. Cancer Cell 2024, 42, 513–534. [Google Scholar] [CrossRef]
  105. Stockwell, B.R.; Friedmann Angeli, J.P.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascón, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 2017, 171, 273–285. [Google Scholar] [CrossRef]
  106. Xu, G.; Wang, H.; Li, X.; Huang, R.; Luo, L. Recent Progress on Targeting Ferroptosis for Cancer Therapy. Biochem. Pharmacol. 2021, 190, 114584. [Google Scholar] [CrossRef]
  107. Luo, L.; Wang, H.; Tian, W.; Zeng, J.; Huang, Y.; Luo, H. Targeting Ferroptosis for Cancer Therapy: Iron Metabolism and Anticancer Immunity. Am. J. Cancer Res. 2021, 11, 5508–5525. [Google Scholar]
  108. Ye, L.; Wen, X.; Qin, J.; Zhang, X.; Wang, Y.; Wang, Z.; Zhou, T.; Di, Y.; He, W. Metabolism-Regulated Ferroptosis in Cancer Progression and Therapy. Cell Death Dis. 2024, 15, 196. [Google Scholar] [CrossRef]
  109. Zhang, W.; Liu, Y.; Liao, Y.; Zhu, C.; Zou, Z. GPX4, Ferroptosis, and Diseases. Biomed. Pharmacother. 2024, 174, 116512. [Google Scholar] [CrossRef]
  110. Ding, K.; Liu, C.; Li, L.; Yang, M.; Jiang, N.; Luo, S.; Sun, L. Acyl-CoA Synthetase ACSL4: An Essential Target in Ferroptosis and Fatty Acid Metabolism. Chin. Med. J. 2023, 136, 2521–2537. [Google Scholar]
  111. Sha, R.; Xu, Y.; Yuan, C.; Sheng, X.; Wu, Z.; Peng, J.; Wang, Y.; Lin, Y.; Zhou, L.; Xu, S.; et al. Predictive and Prognostic Impact of Ferroptosis-Related Genes ACSL4 and GPX4 on Breast Cancer Treated with Neoadjuvant Chemotherapy. EBioMedicine 2021, 71, 103560. [Google Scholar] [CrossRef] [PubMed]
  112. Kong, Y.; Li, J.; Lin, R.; Lu, S.; Rong, L.; Xue, Y.; Fang, Y. Understanding the Unique Mechanism of Ferroptosis: A Promising Therapeutic Target. Front. Cell Dev. Biol. 2024, 11, 1329147. [Google Scholar] [CrossRef] [PubMed]
  113. Zhou, Q.; Meng, Y.; Li, D.; Yao, L.; Le, J.; Liu, Y.; Sun, Y.; Zeng, F.; Chen, X.; Deng, G. Ferroptosis in Cancer: From Molecular Mechanisms to Therapeutic Strategies. Signal Transduct. Target. Ther. 2024, 9, 55. [Google Scholar] [CrossRef]
  114. Shah, R.; Shchepinov, M.S.; Pratt, D.A. Resolving the Role of Lipoxygenases in the Initiation and Execution of Ferroptosis. ACS Cent. Sci. 2018, 4, 387–396. [Google Scholar] [CrossRef]
  115. Ma, T.; Du, J.; Zhang, Y.; Wang, Y.; Wang, B.; Zhang, T. GPX4-Independent Ferroptosis—A New Strategy in Disease’s Therapy. Cell Death Discov. 2022, 8, 434. [Google Scholar] [CrossRef]
  116. Bayır, H.; Anthonymuthu, T.S.; Tyurina, Y.Y.; Patel, S.J.; Amoscato, A.A.; Lamade, A.M.; Yang, Q.; Vladimirov, G.K.; Philpott, C.C.; Kagan, V.E. Achieving Life through Death: Redox Biology of Lipid Peroxidation in Ferroptosis. Cell Chem. Biol. 2020, 27, 387–408. [Google Scholar] [CrossRef]
  117. Lei, G.; Zhuang, L.; Gan, B. Targeting Ferroptosis as a Vulnerability in Cancer. Nat. Rev. Cancer 2022, 22, 381–396. [Google Scholar] [CrossRef]
  118. Conrad, M.; Pratt, D.A. The Chemical Basis of Ferroptosis. Nat. Chem. Biol. 2019, 15, 1137–1147. [Google Scholar] [CrossRef] [PubMed]
  119. Gaschler, M.M.; Stockwell, B.R. Lipid Peroxidation in Cell Death. Biochem. Biophys. Res. Commun. 2017, 482, 419–425. [Google Scholar] [CrossRef]
  120. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial Reactive Oxygen Species (ROS) and ROS-Induced ROS Release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef]
  121. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J., 3rd; Kang, R.; Tang, D. Autophagy Promotes Ferroptosis by Degradation of Ferritin. Autophagy 2016, 12, 1425–1428. [Google Scholar] [CrossRef]
  122. Yang, W.S.; Kim, K.J.; Gaschler, M.M.; Patel, M.; Shchepinov, M.S.; Stockwell, B.R. Peroxidation of Polyunsaturated Fatty Acids by Lipoxygenases Drives Ferroptosis. Proc. Natl. Acad. Sci. USA 2016, 113, E4966–E4975. [Google Scholar] [CrossRef] [PubMed]
  123. Li, F.J.; Long, H.Z.; Zhou, Z.W.; Luo, H.Y.; Xu, S.G.; Gao, L.C. System Xc-/GSH/GPX4 Axis: An Important Antioxidant System for the Ferroptosis in Drug-Resistant Solid Tumor Therapy. Front. Pharmacol. 2022, 13, 910292. [Google Scholar] [CrossRef]
  124. Jiang, X.; Stockwell, B.R.; Conrad, M. Ferroptosis: Mechanisms, Biology and Role in Disease. Nat. Rev. Mol. Cell Biol. 2021, 22, 266–282. [Google Scholar] [CrossRef] [PubMed]
  125. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular Mechanisms and Health Implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef] [PubMed]
  126. Wang, Y.; Yu, G.; Chen, X. Mechanism of Ferroptosis Resistance in Cancer Cells. Cancer Drug Resist. 2024, 7, 47. [Google Scholar] [CrossRef]
  127. Li, J.; Cao, F.; Yin, H.L.; Huang, Z.J.; Lin, Z.T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, Present and Future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef]
  128. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory Responses and Inflammation Associated Diseases in Organs. Oncotarget 2017, 9, 7204–7218. [Google Scholar] [CrossRef]
  129. Raju, M.V.; Chandrasekaran, M.K.; Ahalliya, R.M.; Kanniappan, G.V. Reconnoitering the Role of Lipid Metabolites in Ferroptosis. Adv. Redox Res. 2024, 14, 100117. [Google Scholar]
  130. Yu, Y.; Yan, Y.; Niu, F.; Wang, Y.; Chen, X.; Su, G.; Liu, Y.; Zhao, X.; Qian, L.; Liu, P.; et al. Ferroptosis: A Cell Death Connecting Oxidative Stress, Inflammation and Cardiovascular Diseases. Cell Death Discov. 2021, 7, 193. [Google Scholar] [CrossRef]
  131. Villalón García, I.; Povea Cabello, S.; Álvarez Córdoba, M.; Talaverón Rey, M.; Suárez Rivero, J.M.; Suárez Carrillo, A.; Munuera Cabeza, M.; Reche López, D.; Cilleros Holgado, P.; Piñero Pérez, R.; et al. Vicious Cycle of Lipid Peroxidation and Iron Accumulation in Neurodegeneration. Neural Regen. Res. 2023, 18, 1196–1202. [Google Scholar] [PubMed]
  132. Su, L.J.; Zhang, J.H.; Gomez, H.; Murugan, R.; Hong, X.; Xu, D.; Jiang, F.; Peng, Z.Y. Reactive Oxygen Species Induced Lipid Peroxidation in Apoptosis, Autophagy, and Ferroptosis. Oxidative Med. Cell. Longev. 2019, 2019, 5080843. [Google Scholar] [CrossRef]
  133. Feng, F.; He, S.; Li, X.; He, J.; Luo, L. Mitochondria Mediated Ferroptosis in Diseases Therapy: From Molecular Mechanisms to Implications. Aging Dis. 2024, 15, 714–738. [Google Scholar] [CrossRef]
  134. Napolitano, G.; Fasciolo, G.; Venditti, P. Mitochondrial Management of Reactive Oxygen Species. Antioxidants 2021, 10, 1824. [Google Scholar] [CrossRef]
  135. Li, W.; Liang, L.; Liu, S.; Yi, H.; Zhou, Y. FSP1: A Key Regulator of Ferroptosis. Trends Mol. Med. 2023, 29, 753–764. [Google Scholar] [CrossRef] [PubMed]
  136. Doll, S.; Freitas, F.P.; Shah, R.; Aldrovandi, M.; da Silva, M.C.; Ingold, I.; Goya Grocin, A.; Xavier da Silva, T.N.; Panzilius, E.; Scheel, C.H.; et al. FSP1 Is a Glutathione-Independent Ferroptosis Suppressor. Nature 2019, 575, 693–698. [Google Scholar] [CrossRef] [PubMed]
  137. Cipriano, A.; Viviano, M.; Feoli, A.; Milite, C.; Sarno, G.; Castellano, S.; Sbardella, G. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J. Med. Chem. 2023, 66, 11632–11655. [Google Scholar] [CrossRef]
  138. Endale, H.T.; Tesfaye, W.; Mengstie, T.A. ROS Induced Lipid Peroxidation and Their Role in Ferroptosis. Front. Cell Dev. Biol. 2023, 11, 1226044. [Google Scholar] [CrossRef]
  139. Yang, W.H.; Huang, Z.; Wu, J.; Ding, C.C.; Murphy, S.K.; Chi, J.T. A TAZ-ANGPTL4-NOX2 Axis Regulates Ferroptotic Cell Death and Chemoresistance in Epithelial Ovarian Cancer. Mol. Cancer Res. 2020, 18, 79–90. [Google Scholar] [CrossRef]
  140. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef]
  141. Ru, Q.; Li, Y.; Chen, L.; Wu, Y.; Min, J.; Wang, F. Iron homeostasis and ferroptosis in human diseases: Mechanisms and therapeutic prospects. Signal Transduct. Target. Ther. 2024, 9, 271. [Google Scholar]
  142. Roemhild, K.; von Maltzahn, F.; Weiskirchen, R.; Knüchel, R.; von Stillfried, S.; Lammers, T. Iron metabolism: Pathophysiology and pharmacology. Trends Pharmacol. Sci. 2021, 42, 640–656. [Google Scholar] [CrossRef] [PubMed]
  143. Salimi, Z.; Afsharinasab, M.; Rostami, M.; Eshaghi Milasi, Y.; Mousavi Ezmareh, S.F.; Sakhaei, F.; Mohammad-Sadeghipour, M.; Rasooli Manesh, S.M.; Asemi, Z. Iron chelators: As therapeutic agents in diseases. Ann. Med. Surg. 2024, 86, 2759–2776. [Google Scholar] [CrossRef] [PubMed]
  144. Sato, M.; Kusumi, R.; Hamashima, S.; Kobayashi, S.; Sasaki, S.; Komiyama, Y.; Izumikawa, T.; Conrad, M.; Bannai, S.; Sato, H. The ferroptosis inducer erastin irreversibly inhibits system xc- and synergizes with cisplatin to increase cisplatin’s cytotoxicity in cancer cells. Sci. Rep. 2018, 8, 968. [Google Scholar] [CrossRef] [PubMed]
  145. Chen, H.; Wang, C.; Liu, Z.; He, X.; Tang, W.; He, L.; Feng, Y.; Liu, D.; Yin, Y.; Li, T. Ferroptosis and its multifaceted role in cancer: Mechanisms and therapeutic approach. Antioxidants 2022, 11, 1504. [Google Scholar] [CrossRef]
  146. Weaver, K.; Skouta, R. The selenoprotein glutathione peroxidase 4: From molecular mechanisms to novel therapeutic opportunities. Biomedicines 2022, 10, 891. [Google Scholar] [CrossRef]
  147. Beretta, G.L.; Zaffaroni, N. Radiotherapy-induced ferroptosis for cancer treatment. Front. Mol. Biosci. 2023, 10, 1216733. [Google Scholar] [CrossRef]
  148. Zheng, X.; Jin, X.; Ye, F.; Liu, X.; Yu, B.; Li, Z.; Zhao, T.; Chen, W.; Liu, X.; Di, C.; et al. Ferroptosis: A novel regulated cell death participating in cellular stress response, radiotherapy, and immunotherapy. Exp. Hematol. Oncol. 2023, 12, 65. [Google Scholar] [CrossRef]
  149. Ye, L.F.; Chaudhary, K.R.; Zandkarimi, F.; Harken, A.D.; Kinslow, C.J.; Upadhyayula, P.S.; Dovas, A.; Higgins, D.M.; Tan, H.; Zhang, Y.; et al. Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem. Biol. 2020, 15, 469–484. [Google Scholar] [CrossRef]
  150. Li, L.; Huang, Y.; Jin, X.; Wang, Q.; Su, J.; Guo, L. Dual ratio and ultraprecision quantification of mitochondrial viscosity in ferroptosis enabled by a vibration-based triple-emission fluorescent probe. Anal. Chem. 2023, 95, 17003–17010. [Google Scholar] [CrossRef]
  151. Wu, S.; Yan, Y.; Hou, H.; Huang, Z.; Li, D.; Zhang, X.; Xiao, Y. Polarity-sensitive and membrane-specific probe quantitatively monitoring ferroptosis through fluorescence lifetime imaging. Anal. Chem. 2022, 94, 11238–11247. [Google Scholar] [CrossRef] [PubMed]
  152. Chen, X.; Comish, P.B.; Tang, D.; Kang, R. Characteristics and biomarkers of ferroptosis. Front. Cell Dev. Biol. 2021, 9, 637162. [Google Scholar] [CrossRef] [PubMed]
  153. Houglum, K.; Filip, M.; Witztum, J.L.; Chojkier, M. Malondialdehyde and 4-hydroxynonenal protein adducts in plasma and liver of rats with iron overload. J. Clin. Investig. 1990, 86, 1991–1998. [Google Scholar] [CrossRef]
  154. Feng, H.; Schorpp, K.; Jin, J.; Yozwiak, C.E.; Hoffstrom, B.G.; Decker, A.M.; Rajbhandari, P.; Stokes, M.E.; Bender, H.G.; Csuka, J.M.; et al. Transferrin receptor is a specific ferroptosis marker. Cell Rep. 2020, 30, 3411–3423.e7. [Google Scholar] [CrossRef]
  155. Koppula, P.; Zhuang, L.; Gan, B. Cystine transporter SLC7A11/xCT in cancer: Ferroptosis, nutrient dependency, and cancer therapy. Protein Cell 2021, 12, 599–620. [Google Scholar] [CrossRef] [PubMed]
  156. Soga, T.; Baran, R.; Suematsu, M.; Ueno, Y.; Ikeda, S.; Sakurakawa, T.; Kakazu, Y.; Ishikawa, T.; Robert, M.; Nishioka, T.; et al. Differential metabolomics reveals ophthalmic acid as an oxidative stress biomarker indicating hepatic glutathione consumption. J. Biol. Chem. 2006, 281, 16768–16776. [Google Scholar] [CrossRef]
  157. Kirkwood-Donelson, K.I.; Jarmusch, A.K.; Bortner, C.D.; Merrick, B.A.; Sinha, B.K. Metabolic consequences of erastin-induced ferroptosis in human ovarian cancer cells: An untargeted metabolomics study. Front. Mol. Biosci. 2025, 11, 1520876. [Google Scholar] [CrossRef]
  158. Sinha, B.K.; Bortner, C.D.; Jarmusch, A.K.; Tokar, E.J.; Murphy, C.; Wu, X.; Winter, H.; Cannon, R.E. Ferroptosis-mediated cell death induced by NCX4040, the non-steroidal nitric oxide donor, in human colorectal cancer cells: Implications in therapy. Cells 2023, 12, 1626. [Google Scholar] [CrossRef]
  159. Sun, J.; Ren, H.; Wang, J.; Xiao, X.; Zhu, L.; Wang, Y.; Yang, L. CHAC1: A master regulator of oxidative stress and ferroptosis in human diseases and cancers. Front. Cell Dev. Biol. 2024, 12, 1458716. [Google Scholar] [CrossRef]
  160. Sinha, B.K.; Murphy, C.; Brown, S.M.; Silver, B.B.; Tokar, E.J.; Bortner, C.D. Mechanisms of cell death induced by erastin in human ovarian tumor cells. Int. J. Mol. Sci. 2024, 25, 8666. [Google Scholar] [CrossRef]
  161. Qi, Y.L.; Wang, H.R.; Chen, L.L.; Duan, Y.T.; Yang, S.Y.; Zhu, H.L. Recent advances in small-molecule fluorescent probes for studying ferroptosis. Chem. Soc. Rev. 2022, 51, 7752–7778. [Google Scholar] [CrossRef]
  162. Yin, J.; Zhan, J.; Hu, Q.; Huang, S.; Lin, W. Fluorescent probes for ferroptosis bioimaging: Advances, challenges, and prospects. Chem. Soc. Rev. 2023, 52, 2011–2030. [Google Scholar] [CrossRef]
  163. Tian, X.; Cheng, J.; Yang, L.; Li, Z.; Yu, M. A NIR dual-channel fluorescent probe for fluctuations of intracellular polarity and H2O2 and its applications for the visualization of inflammation and ferroptosis. Chem. Biomed. Imaging 2024, 2, 518–525. [Google Scholar] [CrossRef] [PubMed]
  164. Nizami, Z.N.; Aburawi, H.E.; Semlali, A.; Muhammad, K.; Iratni, R. Oxidative stress inducers in cancer therapy: Preclinical and clinical evidence. Antioxidants 2023, 12, 1159. [Google Scholar] [CrossRef]
  165. Liang, Z.; Zhao, W.; Li, X.; Wang, L.; Meng, L.; Yu, R. Cisplatin synergizes with PRLX93936 to induce ferroptosis in non-small cell lung cancer cells. Biochem. Biophys. Res. Commun. 2021, 569, 79–85. [Google Scholar] [CrossRef] [PubMed]
  166. Gaschler, M.M.; Andia, A.A.; Liu, H.; Csuka, J.M.; Hurlocker, B.; Vaiana, C.A.; Heindel, D.W.; Zuckerman, D.S.; Bos, P.H.; Reznik, E.; et al. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat. Chem. Biol. 2018, 14, 507–515. [Google Scholar] [CrossRef] [PubMed]
  167. Perera, L.; Brown, S.M.; Silver, B.B.; Tokar, E.J.; Sinha, B.K. Ferroptosis inducers erastin and RSL3 enhance adriamycin and topotecan sensitivity in ABCB1/ABCG2-expressing tumor cells. Int. J. Mol. Sci. 2025, 26, 635. [Google Scholar] [CrossRef]
  168. Yang, X.; Zhang, M.; Xia, W.; Mai, Z.; Ye, Y.; Zhao, B.; Song, Y. CHAC1 promotes cell ferroptosis and enhances radiation sensitivity in thyroid carcinoma. Neoplasma 2023, 70, 777–786. [Google Scholar] [CrossRef]
  169. Cheff, D.M.; Huang, C.; Scholzen, K.C.; Gencheva, R.; Ronzetti, M.H.; Cheng, Q.; Hall, M.D.; Arnér, E.S.J. The ferroptosis inducing compounds RSL3 and ML162 are not direct inhibitors of GPX4 but of TXNRD1. Redox Biol. 2023, 62, 102703. [Google Scholar] [CrossRef]
  170. Desterke, C.; Xiang, Y.; Elhage, R.; Duruel, C.; Chang, Y.; Hamaï, A. Ferroptosis inducers upregulate PD-L1 in recurrent triple-negative breast cancer. Cancers 2023, 16, 155. [Google Scholar] [CrossRef]
  171. Song, X.; Wang, X.; Liu, Z.; Yu, Z. Role of GPX4-mediated ferroptosis in the sensitivity of triple-negative breast cancer cells to gefitinib. Front. Oncol. 2020, 10, 597434. [Google Scholar] [CrossRef] [PubMed]
  172. Ma, B.; Zhong, Y.; Chen, R.; Zhan, X.; Huang, G.; Xiong, Y.; Tan, B. Tripterygium glycosides reverse chemotherapy resistance in ovarian cancer by targeting the NRF2/GPX4 signal axis to induce ferroptosis of drug-resistant human epithelial ovarian cancer cells. Biochem. Biophys. Res. Commun. 2023, 665, 178–186. [Google Scholar] [CrossRef] [PubMed]
  173. Li, N.; Jiang, X.; Zhang, Q.; Huang, Y.; Wei, J.; Zhang, H.; Luo, H. Synergistic suppression of ovarian cancer by combining NRF2 and GPX4 inhibitors: In vitro and in vivo evidence. J. Ovarian Res. 2024, 17, 49. [Google Scholar] [CrossRef]
  174. Yan, G.; Huang, H.; Lu, Z.; Chen, M.; Wang, X.; Zhong, P.; Qin, C.; Mo, S.; Han, C.; Luo, X.; et al. Comprehensive pan-cancer analysis and functional studies reveal SLC2A6 as a ferroptosis modulator in hepatocellular carcinoma. Sci. Rep. 2025, 15, 2545. [Google Scholar] [CrossRef] [PubMed]
  175. Chen, Y.S.; Lee, C.H.; Hsieh, Y.H.; Chiou, H.L.; Hung, M.C.; Lee, H.L. Sorafenib, a tyrosine kinase inhibitor, synergistically enhances the ferroptosis effects of asiatic acid in hepatocellular carcinoma cells. Environ. Toxicol. 2025, 40, 79–87. [Google Scholar] [CrossRef]
  176. Cui, Y.; Li, Y.; Xu, Y.; Liu, X.; Kang, X.; Zhu, J.; Long, S.; Han, Y.; Xue, C.; Sun, Z.; et al. SLC7A11 protects luminal A breast cancer cells against ferroptosis induced by CDK4/6 inhibitors. Redox Biol. 2024, 76, 103304. [Google Scholar] [CrossRef]
  177. Skeie, B.S.; Bragstad, S.; Sarowar, S.; Behbahani, M.; Filippi, C.; Knisely, J.; Schulder, M.; Goplen, D.; Eide, G.E.; Heggdal, J.I.; et al. CTNI-40. Phase I Trial of Sulfasalazine Combined with Stereotactic Radiosurgery or Recurrent Glioblastoma: Study Protocol for NCT04205357. Neuro Oncol. 2022, 24, vii80–vii81. [Google Scholar] [CrossRef]
  178. ClinicalTrials.gov ID NCT06048367. Carbon Nanoparticle-Loaded Iron [CNSI-Fe(II)] in the Treatment of Advanced Solid Tumor (CNSI-Fe(II)). Available online: https://clinicaltrials.gov/ (accessed on 11 February 2025).
Figure 1. A flow model showing the steps in ferroptosis involving various membrane and intracellular components, including transferrin receptors, glutamine-cystine antiporter (Xc), LOX, GPX, GSH, iron, and lipid peroxides [83].
Figure 1. A flow model showing the steps in ferroptosis involving various membrane and intracellular components, including transferrin receptors, glutamine-cystine antiporter (Xc), LOX, GPX, GSH, iron, and lipid peroxides [83].
Ijms 26 03852 g001
Figure 2. A summary of the various molecular mechanisms of ferroptosis, with key genes involved in the induction of ferroptotic cell death in cancer cells.
Figure 2. A summary of the various molecular mechanisms of ferroptosis, with key genes involved in the induction of ferroptotic cell death in cancer cells.
Ijms 26 03852 g002
Figure 3. A summary of various types of ferroptosis and their key genes that contribute to ferroptosis cell death [121,122,123,124].
Figure 3. A summary of various types of ferroptosis and their key genes that contribute to ferroptosis cell death [121,122,123,124].
Ijms 26 03852 g003
Table 1. Various mechanisms of ferroptosis involve molecular genes, proteins, and therapeutic implications for cancer.
Table 1. Various mechanisms of ferroptosis involve molecular genes, proteins, and therapeutic implications for cancer.
MechanismMolecular Genes/ProteinsPathwaysTherapeutic ImplicationsReferences
AutophagyATG5, ATG7, LC3BAutophagic degradationAutophagy inhibition[38]
p53 PathwayTP53, MDM2Tumor suppressor regulationp53-reactivating therapies[45]
NRF2 PathwayNFE2L2, KEAP1Antioxidant response element (ARE) regulationNRF2 activation[113]
Redox ImbalanceGSHReactive oxygen species (ROS) regulationAntioxidant therapies[123]
InflammationTNF, IL-1β, NF-κBCytokine signalingAnti-inflammatory therapies[130,131]
Mitochondrial DysfunctionBCL-2Mitochondrial outer membrane permeabilizationMitochondria-targeting therapies[132,133]
Lipid PeroxidationGPX4, ACSL4, SLC7A11Fatty acid synthesis, antioxidant defensesTargeting lipid metabolism[134,135,136,137,138]
Iron MetabolismNCOA4, TFRC, SLC40A1Ferritinophagy, iron uptakeIron chelation therapy[139,140,141]
Table 2. Various types of ferroptosis with molecular descriptions, involved molecular genes, and associated cancer hallmarks.
Table 2. Various types of ferroptosis with molecular descriptions, involved molecular genes, and associated cancer hallmarks.
TypesMolecular MechanismsKey Genes InvolvedRelevant Cancer HallmarksReferences
Oxidative FerroptosisAccumulation of oxygen species (ROS) and mitochondrial damageP53, KEAP1, NFE2L2Cancer cell sensitivity[100]
Autophagy-Dependent FerroptosisInvolves autophagic degradation of damaged cellular componentsATG5, ATG7, LC3BCancer cell survival[121]
Classical FerroptosisRegulated cell death driven by iron-dependent lipid peroxidationACSL4, GPX4, SLC7A11Cancer cell vulnerability[124,125]
Ferroptosis-like Cell DeathShares features with ferroptosis but lacks lipid peroxidationALOX12Cancer therapy potential[127]
Mitochondrial FerroptosisMitochondrial dysfunction, including changes in membrane potentialBCL-2, BAX, BakCancer therapy target[133]
Non-Canonical FerroptosisIndependent of GPX4 NCOA4, TFRC, SLC40A1Resistance in cancer cells[135,136]
Iron Overload-Induced FerroptosisFenton reaction and lipid peroxidationFerritinResistance in cancer cells[141]
Erastin-Induced FerroptosisSystem Xc−, an antiporter of cystine/glutamateGPX4Cancer therapy target[144,145]
Radiation-Induced FerroptosisRadiation leads to the generation of ROSGPX4Cancer cell sensitivity[147,148]
Table 3. Preclinical and clinical evidence on modulators of ferroptosis in combinatorial anticancer drug approaches, with details on various molecular targets.
Table 3. Preclinical and clinical evidence on modulators of ferroptosis in combinatorial anticancer drug approaches, with details on various molecular targets.
Inducers of FerroptosisMolecular Target/
Pathway
Combinatorial Anticancer Drug ApproachPreclinical/Clinical EvidenceReferences
SeleniteGPX4/SELENOPAnticancer drugEnhanced antitumor effect in ovarian cancer via induction of ferroptosis and inhibition of GPX4-mediated antioxidant defenses[143]
PRLX93936 inhibitor of GPX4GPX4Cisplatinupregulation of ROS, lipid peroxidation, and Fe2+[165]
FINO2GPX4/SLC7A11Anticancer drugFINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation[166]
ML162, ML210Selenoprotein, TXNRD1Anticancer drugInhibition of selenoprotein, TXNRD1[169]
Lentivirus sh-GPX4GPX4GefitinibIncreased antitumor efficacy in breast cancer via inhibition of GPX4-mediated ferroptosis and induction of SELENOP-mediated selenium depletion[171]
Tripterygium glycosidesGPX4/NRF2CisplatinSynergistic antitumor effect in ovarian cancer via induction of ferroptosis and inhibition of NRF2-mediated GPX4 expression[172]
ML385GPX4/NRF2Anticancer drugsEnhanced antitumor efficacy in ovarian cancer via inhibition of NRF2-mediated GPX4 expression and induction of ferroptosis[173]
GPX4 inhibitor (RSL3)GPX4/NRF2SorafenibSynergistic antitumor effect in HCC via inhibition of NRF2-mediated GPX4 expression and induction of ferroptosis[175]
FINO2GPX4/SLC7A11PaclitaxelIncreased antitumor efficacy in breast cancer via inhibition of GPX4-mediated ferroptosis and induction of SLC7A11-mediated glutathione depletion[176]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Singh, M.; Arora, H.L.; Naik, R.; Joshi, S.; Sonawane, K.; Sharma, N.K.; Sinha, B.K. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. Int. J. Mol. Sci. 2025, 26, 3852. https://doi.org/10.3390/ijms26083852

AMA Style

Singh M, Arora HL, Naik R, Joshi S, Sonawane K, Sharma NK, Sinha BK. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. International Journal of Molecular Sciences. 2025; 26(8):3852. https://doi.org/10.3390/ijms26083852

Chicago/Turabian Style

Singh, Mansaa, Hasmiq L. Arora, Rutuja Naik, Shravani Joshi, Kaveri Sonawane, Nilesh Kumar Sharma, and Birandra K. Sinha. 2025. "Ferroptosis in Cancer: Mechanism and Therapeutic Potential" International Journal of Molecular Sciences 26, no. 8: 3852. https://doi.org/10.3390/ijms26083852

APA Style

Singh, M., Arora, H. L., Naik, R., Joshi, S., Sonawane, K., Sharma, N. K., & Sinha, B. K. (2025). Ferroptosis in Cancer: Mechanism and Therapeutic Potential. International Journal of Molecular Sciences, 26(8), 3852. https://doi.org/10.3390/ijms26083852

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop